Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 17(5): e1009593, 2021 05.
Article in English | MEDLINE | ID: mdl-34015056

ABSTRACT

Type I interferons (IFN-I) are antiviral cytokines that signal through the ubiquitous IFN-I receptor (IFNAR). Following footpad infection with ectromelia virus (ECTV), a mouse-specific pathogen, C57BL/6 (B6) mice survive without disease, while B6 mice broadly deficient in IFNAR succumb rapidly. We now show that for survival to ECTV, only hematopoietic cells require IFNAR expression. Survival to ECTV specifically requires IFNAR in both natural killer (NK) cells and monocytes. However, intrinsic IFNAR signaling is not essential for adaptive immune cell responses or to directly protect non-hematopoietic cells such as hepatocytes, which are principal ECTV targets. Mechanistically, IFNAR-deficient NK cells have reduced cytolytic function, while lack of IFNAR in monocytes dampens IFN-I production and hastens virus dissemination. Thus, during a pathogenic viral infection, IFN-I coordinates innate immunity by stimulating monocytes in a positive feedback loop and by inducing NK cell cytolytic function.


Subject(s)
Ectromelia virus/immunology , Ectromelia, Infectious/immunology , Receptor, Interferon alpha-beta/metabolism , Signal Transduction , Animals , Cytokines/immunology , Disease Resistance , Ectromelia, Infectious/virology , Female , Hepatocytes/immunology , Hepatocytes/virology , Immunity, Innate , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Male , Mice , Mice, Inbred C57BL , Monocytes/immunology , Monocytes/virology , Receptor, Interferon alpha-beta/genetics
2.
Mol Ther ; 29(9): 2769-2781, 2021 09 01.
Article in English | MEDLINE | ID: mdl-33992803

ABSTRACT

It is well established that memory CD8 T cells protect susceptible strains of mice from mousepox, a lethal viral disease caused by ectromelia virus (ECTV), the murine counterpart to human variola virus. While mRNA vaccines induce protective antibody (Ab) responses, it is unknown whether they also induce protective memory CD8 T cells. We now show that immunization with different doses of unmodified or N(1)-methylpseudouridine-modified mRNA (modified mRNA) in lipid nanoparticles (LNP) encoding the ECTV gene EVM158 induced similarly strong CD8 T cell responses to the epitope TSYKFESV, albeit unmodified mRNA-LNP had adverse effects at the inoculation site. A single immunization with 10 µg modified mRNA-LNP protected most susceptible mice from mousepox, and booster vaccination increased the memory CD8 T cell pool, providing full protection. Moreover, modified mRNA-LNP encoding TSYKFESV appended to green fluorescent protein (GFP) protected against wild-type ECTV infection while lymphocytic choriomeningitis virus glycoprotein (GP) modified mRNA-LNP protected against ECTV expressing GP epitopes. Thus, modified mRNA-LNP can be used to create protective CD8 T cell-based vaccines against viral infections.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Ectromelia virus/immunology , Ectromelia, Infectious/prevention & control , Viral Proteins/genetics , mRNA Vaccines/administration & dosage , Animals , Drug Compounding , Ectromelia, Infectious/immunology , Immunization, Secondary , Immunologic Memory , Liposomes , Male , Mice , Nanoparticles , Peptides/chemistry , Peptides/genetics , Peptides/immunology , Pseudouridine/analogs & derivatives , Pseudouridine/chemistry , Viral Proteins/chemistry , Viral Proteins/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/chemistry , Viral Vaccines/pharmacology , mRNA Vaccines/chemistry , mRNA Vaccines/pharmacology
3.
J Exp Med ; 218(5)2021 05 03.
Article in English | MEDLINE | ID: mdl-33765134

ABSTRACT

Natural killer (NK) cell activation depends on the signaling balance of activating and inhibitory receptors. CD94 forms inhibitory receptors with NKG2A and activating receptors with NKG2E or NKG2C. We previously demonstrated that CD94-NKG2 on NK cells and its ligand Qa-1b are important for the resistance of C57BL/6 mice to lethal ectromelia virus (ECTV) infection. We now show that NKG2C or NKG2E deficiency does not increase susceptibility to lethal ECTV infection, but overexpression of Qa-1b in infected cells does. We also demonstrate that Qa-1b is down-regulated in infected and up-regulated in bystander inflammatory monocytes and B cells. Moreover, NK cells activated by ECTV infection kill Qa-1b-deficient cells in vitro and in vivo. Thus, during viral infection, recognition of Qa-1b by activating CD94/NKG2 receptors is not critical. Instead, the levels of Qa-1b expression are down-regulated in infected cells but increased in some bystander immune cells to respectively promote or inhibit their killing by activated NK cells.


Subject(s)
B-Lymphocytes/immunology , Cytotoxicity, Immunologic/immunology , Ectromelia virus/immunology , Histocompatibility Antigens Class I/immunology , Killer Cells, Natural/immunology , Virus Diseases/immunology , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Bystander Effect/immunology , Cytotoxicity, Immunologic/genetics , Ectromelia virus/physiology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Killer Cells, Natural/metabolism , Killer Cells, Natural/virology , Male , Mice, Inbred C57BL , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily C/genetics , NK Cell Lectin-Like Receptor Subfamily C/immunology , NK Cell Lectin-Like Receptor Subfamily C/metabolism , NK Cell Lectin-Like Receptor Subfamily D/genetics , NK Cell Lectin-Like Receptor Subfamily D/immunology , NK Cell Lectin-Like Receptor Subfamily D/metabolism , Virus Diseases/virology
5.
Aging Cell ; 19(7): e13170, 2020 07.
Article in English | MEDLINE | ID: mdl-32657004

ABSTRACT

It is known that aging decreases natural resistance to viral diseases due to dysfunctional innate and adaptive immune responses, but the nature of these dysfunctions, particularly in regard to innate immunity, is not well understood. We have previously shown that C57BL/6J (B6) mice lose their natural resistance to footpad infection with ectromelia virus (ECTV) due to impaired maturation and recruitment of natural killer (NK) cells to the draining popliteal lymph node (dLN). More recently, we have also shown that in young B6 mice infected with ECTV, the recruitment of NK cells is dependent on a complex cascade whereby migratory dendritic cells (mDCs) traffic from the skin to the dLN, where they produce CCL2 and CCL7 to recruit inflammatory monocytes (iMOs). In the dLN, mDCs also upregulate NKG2D ligands to induce interferon gamma (IFN-γ) expression by group 1 innate lymphoid cells (G1-ILCs), mostly NK in cells but also some ILC1. In response to the IFN-γ, the incoming uninfected iMOs secret CXCL9 to recruit the critical NK cells. Here, we show that in aged B6 mice, the trafficking of mDCs to the dLN in response to ECTV is decreased, resulting in impaired IFN-γ expression by G1-ILCs, reduced accumulation of iMOs, and attenuated CXCL9 production by iMOs, which likely contributes to decrease in NK cell recruitment. Together, these data indicate that defects in the mDC response to viral infection during aging result in a reduced innate immune response in the dLN and contribute to increased susceptibility to viral disease in the aged.


Subject(s)
Dendritic Cells/metabolism , Ectromelia virus/immunology , Immunity, Innate/immunology , Lymph Nodes/metabolism , Aging , Animals , Mice
6.
J Immunol ; 204(11): 2961-2972, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32284333

ABSTRACT

CMV has been proposed to play a role in cancer progression and invasiveness. However, CMV has been increasingly studied as a cancer vaccine vector, and multiple groups, including ours, have reported that the virus can drive antitumor immunity in certain models. Our previous work revealed that intratumoral injections of wild-type murine CMV (MCMV) into B16-F0 melanomas caused tumor growth delay in part by using a viral chemokine to recruit macrophages that were subsequently infected. We now show that MCMV acts as a STING agonist in the tumor. MCMV infection of tumors in STING-deficient mice resulted in normal recruitment of macrophages to the tumor, but poor recruitment of CD8+ T cells, reduced production of inflammatory cytokines and chemokines, and no delay in tumor growth. In vitro, expression of type I IFN was dependent on both STING and the type I IFNR. Moreover, type I IFN alone was sufficient to induce cytokine and chemokine production by macrophages and B16 tumor cells, suggesting that the major role for STING activation was to produce type I IFN. Critically, viral infection of wild-type macrophages alone was sufficient to restore tumor growth delay in STING-deficient animals. Overall, these data show that MCMV infection and sensing in tumor-associated macrophages through STING signaling is sufficient to promote antitumor immune responses in the B16-F0 melanoma model.


Subject(s)
Herpesviridae Infections/immunology , Melanoma/immunology , Membrane Proteins/metabolism , Muromegalovirus/physiology , Skin Neoplasms/immunology , Tumor-Associated Macrophages/immunology , Animals , Cell Movement , Disease Models, Animal , Humans , Immunity/genetics , Interferon Type I/metabolism , Melanoma/virology , Melanoma, Experimental , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Skin Neoplasms/virology , Tumor Burden , Tumor Microenvironment
7.
J Immunol ; 204(6): 1582-1591, 2020 03 15.
Article in English | MEDLINE | ID: mdl-32015010

ABSTRACT

NK cells play an important role in antiviral resistance. The integrin α2, which dimerizes with integrin ß1, distinguishes NK cells from innate lymphoid cells 1 and other leukocytes. Despite its use as an NK cell marker, little is known about the role of α2ß1 in NK cell biology. In this study, we show that in mice α2ß1 deficiency does not alter the balance of NK cell/ innate lymphoid cell 1 generation and slightly decreases the number of NK cells in the bone marrow and spleen without affecting NK cell maturation. NK cells deficient in α2ß1 had no impairment at entering or distributing within the draining lymph node of ectromelia virus (ECTV)-infected mice or at becoming effectors but proliferated poorly in response to ECTV and did not increase in numbers following infection with mouse CMV (MCMV). Still, α2ß1-deficient NK cells efficiently protected from lethal mousepox and controlled MCMV titers in the spleen. Thus, α2ß1 is required for optimal NK cell proliferation but is dispensable for protection against ECTV and MCMV, two well-established models of viral infection in which NK cells are known to be important.


Subject(s)
Ectromelia, Infectious/immunology , Herpesviridae Infections/immunology , Integrin alpha2beta1/metabolism , Killer Cells, Natural/immunology , Animals , Cell Count , Cell Proliferation , Disease Models, Animal , Ectromelia virus/immunology , Ectromelia, Infectious/blood , Ectromelia, Infectious/virology , Female , Herpesviridae Infections/blood , Herpesviridae Infections/virology , Humans , Immunity, Innate , Integrin alpha2beta1/immunology , Killer Cells, Natural/metabolism , Male , Mice , Muromegalovirus/immunology , Virus Replication/immunology
8.
J Virol ; 94(5)2020 02 14.
Article in English | MEDLINE | ID: mdl-31776282

ABSTRACT

Chronic viral infections. like those of humans with cytomegalovirus, human immunodeficiency virus (even when under antiretroviral therapy), and hepatitis C virus or those of mice with lymphocytic choriomeningitis virus (LCMV) clone 13 (CL13), result in immune dysfunction that predisposes the host to severe infections with unrelated pathogens. It is known that C57BL/6 (B6) mice are resistant to mousepox, a lethal disease caused by the orthopoxvirus ectromelia virus (ECTV), and that this resistance requires natural killer (NK) cells and other immune cells. We show that most B6 mice chronically infected with CL13 succumb to mousepox but that most of those that recovered from acute infection with the LCMV Armstrong (Arm) strain survive. We also show that B6 mice chronically infected with CL13 and those that recovered from Arm infection have a reduced frequency and a reduced number of NK cells. However, at steady state, NK cells in mice that have recovered from Arm infection mature normally and, in response to ECTV, get activated, become more mature, proliferate, and increase their cytotoxicity in vivo Conversely, in mice chronically infected with CL13, NK cells are immature and residually activated, and following ECTV infection, they do not mature, proliferate, or increase their cytotoxicity. Given the well-established importance of NK cells in resistance to mousepox, these data suggest that the NK cell dysfunction caused by CL13 persistence may contribute to the susceptibility of CL13-infected mice to mousepox. Whether chronic infections similarly affect NK cells in humans should be explored.IMPORTANCE Infection of adult mice with the clone 13 (CL13) strain of lymphocytic choriomeningitis virus (LCMV) is extensively used as a model of chronic infection. In this paper, we show that mice chronically infected with CL13 succumb to challenge with ectromelia virus (ECTV; the agent of mousepox) and that natural killer (NK) cells in CL13-infected mice are reduced in numbers and have an immature and partially activated phenotype but do respond to ECTV. These data may provide additional clues why humans chronically infected with certain pathogens are less resistant to viral diseases.


Subject(s)
Ectromelia virus/immunology , Ectromelia, Infectious/immunology , Killer Cells, Natural/immunology , Lymphocytic Choriomeningitis/immunology , ATP Binding Cassette Transporter, Subfamily B, Member 2/genetics , Animals , Cytokines/metabolism , Disease Models, Animal , Female , Lymphocytic choriomeningitis virus/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
9.
J Virol ; 94(5)2020 02 14.
Article in English | MEDLINE | ID: mdl-31826990

ABSTRACT

It is well established that chronic viral infections can cause immune suppression, resulting in increased susceptibility to other infectious diseases. However, the effects of chronic viral infection on T-cell responses and vaccination against highly pathogenic viruses are not well understood. We have recently shown that C57BL/6 (B6) mice lose their natural resistance to wild-type (WT) ectromelia virus (ECTV) when chronically infected with lymphocytic choriomeningitis virus (LCMV) clone 13 (CL13). Here we compared the T-cell response to ECTV in previously immunologically naive mice that were chronically infected with CL13 or that were convalescent from acute infection with the Armstrong (Arm) strain of LCMV. Our results show that mice that were chronically infected with CL13 but not those that had recovered from Arm infection have highly defective ECTV-specific CD8+ and CD4+ T-cell responses to WT ECTV. These defects are at least partly due to the chronic infection environment. In contrast to mice infected with WT ECTV, mice chronically infected with CL13 survived without signs of disease when infected with ECTV-Δ036, a mutant ECTV strain that is highly attenuated. Strikingly, mice chronically infected with CL13 mounted a strong CD8+ T-cell response to ECTV-Δ036 and survived without signs of disease after a subsequent challenge with WT ECTV. Our work suggests that enhanced susceptibility to acute viral infections in chronically infected individuals can be partly due to poor T-cell responses but that sufficient T-cell function can be recovered and resistance to acute infection can be restored by immunization with highly attenuated vaccines.IMPORTANCE Chronic viral infections may result in immunosuppression and enhanced susceptibility to infections with other pathogens. For example, we have recently shown that mice chronically infected with lymphocytic choriomeningitis virus (LCMV) clone 13 (CL13) are highly susceptible to mousepox, a disease that is caused by ectromelia virus and that is the mouse homolog of human smallpox. Here we show chronic CL13 infection severely disrupts the expansion, proliferation, activation, and cytotoxicity of T cells in response due at least in part to the suppressive effects of the chronic infection milieu. Notably, despite this profound immunodeficiency, mice chronically infected with CL13 could be protected by vaccination with a highly attenuated variant of ECTV. These results demonstrate that protective vaccination of immunosuppressed individuals is possible, provided that proper immunization tools are used.


Subject(s)
Ectromelia, Infectious/immunology , Immunity, Innate/immunology , Immunization , Lymphocytic Choriomeningitis/immunology , T-Lymphocytes/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Ectromelia virus/immunology , Female , Humans , Immune Tolerance , Immunologic Memory , Lymphocytic choriomeningitis virus/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Vaccination
10.
PLoS Pathog ; 15(12): e1008239, 2019 12.
Article in English | MEDLINE | ID: mdl-31877196

ABSTRACT

Cells sensing infection produce Type I interferons (IFN-I) to stimulate Interferon Stimulated Genes (ISGs) that confer resistance to viruses. During lympho-hematogenous spread of the mouse pathogen ectromelia virus (ECTV), the adaptor STING and the transcription factor IRF7 are required for IFN-I and ISG induction and resistance to ECTV. However, it is unknown which cells sense ECTV and which pathogen recognition receptor (PRR) upstream of STING is required for IFN-I and ISG induction. We found that cyclic-GMP-AMP (cGAMP) synthase (cGAS), a DNA-sensing PRR, is required in bone marrow-derived (BMD) but not in other cells for IFN-I and ISG induction and for resistance to lethal mousepox. Also, local administration of cGAMP, the product of cGAS that activates STING, rescues cGAS but not IRF7 or IFN-I receptor deficient mice from mousepox. Thus, sensing of infection by BMD cells via cGAS and IRF7 is critical for resistance to a lethal viral disease in a natural host.


Subject(s)
Bone Marrow/virology , Ectromelia virus/pathogenicity , Ectromelia, Infectious/virology , Nucleotides, Cyclic/metabolism , Animals , Bone Marrow/metabolism , Host-Pathogen Interactions/immunology , Immunity, Innate/immunology , Interferon Type I/metabolism , Mice, Transgenic , Nucleotidyltransferases/genetics , Protein Serine-Threonine Kinases/metabolism
11.
Cell Rep ; 29(10): 3047-3059.e3, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31801072

ABSTRACT

During disseminating viral infections, a swift innate immune response (IIR) in the draining lymph node (dLN) that restricts systemic viral spread is critical for optimal resistance to disease. However, it is unclear how this IIR is orchestrated. We show that after footpad infection of mice with ectromelia virus, dendritic cells (DCs) highly expressing major histocompatibility complex class II (MHC class IIhi DCs), including CD207+ epidermal Langerhans cells (LCs), CD103+CD207+ double-positive dermal DCs (DP-DCs), and CD103-CD207- double-negative dermal DCs (DN-DCs) migrate to the dLN from the skin carrying virus. MHC class IIhi DCs, predominantly LCs and DP-DCs, are the first cells upregulating IIR cytokines in the dLN. Preventing MHC class IIhi DC migration or depletion of LCs, but not DP-DC deficiency, suppresses the IIR in the dLN and results in high viral lethality. Therefore, LCs are the architects of an early IIR in the dLN that is critical for optimal resistance to a disseminating viral infection.


Subject(s)
Antiviral Agents/immunology , Immunity, Innate/immunology , Langerhans Cells/immunology , Lymph Nodes/immunology , Animals , Cell Line , Cell Movement/immunology , Cytokines/immunology , Dendritic Cells/immunology , Female , Histocompatibility Antigens Class II/immunology , Male , Mice , Mice, Inbred C57BL , Skin/immunology , Up-Regulation/immunology
12.
Cell Rep ; 24(1): 142-154, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29972776

ABSTRACT

Circulating natural killer (NK) cells help protect the host from lympho-hematogenous acute viral diseases by rapidly entering draining lymph nodes (dLNs) to curb virus dissemination. Here, we identify a highly choreographed mechanism underlying this process. Using footpad infection with ectromelia virus, a pathogenic DNA virus of mice, we show that TLR9/MyD88 sensing induces NKG2D ligands in virus-infected, skin-derived migratory dendritic cells (mDCs) to induce production of IFN-γ by classical NK cells and other types of group 1 innate lymphoid cells (ILCs) already in dLNs, via NKG2D. Uninfected inflammatory monocytes, also recruited to dLNs by mDCs in a TLR9/MyD88-dependent manner, respond to IFN-γ by secreting CXCL9 for optimal CXCR3-dependent recruitment of circulating NK cells. This work unveils a TLR9/MyD88-dependent mechanism whereby in dLNs, three cell types-mDCs, group 1 ILCs (mostly NK cells), and inflammatory monocytes-coordinate the recruitment of protective circulating NK cells to dLNs.


Subject(s)
Cell Movement , Dendritic Cells/immunology , Ectromelia virus/physiology , Inflammation/pathology , Killer Cells, Natural/immunology , Lymph Nodes/virology , Lymphocytes/immunology , Monocytes/immunology , Animals , Chemokine CXCL9/metabolism , Endothelium/virology , Female , Immunity, Innate , Interferons/metabolism , Ligands , Lymph Nodes/immunology , Male , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/metabolism , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Receptors, CXCR3/metabolism , Stromal Cells/metabolism , Toll-Like Receptor 9/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...